51
|
Iacopo G, Allinovi M, Caroti L, Cirami LC. Broad spectrum of interferon-related nephropathies—glomerulonephritis, systemic lupus erythematosus-like syndrome and thrombotic microangiopathy: A case report and review of literature. World J Nephrol 2019. [DOI: 10.5527/wjgo.v8.i7.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
52
|
Gianassi I, Allinovi M, Caroti L, Cirami LC. Broad spectrum of interferon-related nephropathies-glomerulonephritis, systemic lupus erythematosus-like syndrome and thrombotic microangiopathy: A case report and review of literature. World J Nephrol 2019; 8:109-117. [PMID: 31750091 PMCID: PMC6853798 DOI: 10.5527/wjn.v8.i7.109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/04/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Interferons (IFNs) are characterized by a wide range of biological effects, which justifies their potential therapeutic use in several pathologies, but also elicit a wide array of adverse effects in almost every organ system. Among them, renal involvement is probably one of the most complex to identify.
CASE SUMMARY We describe four cases of kidney damage caused by different IFN formulations: IFN-β-related thrombotic microangiopathy, IFN-β-induced systemic lupus erythematosus, and two cases of membranous nephropathy secondary to pegylated-IFN-α 2B. In each case, we carefully excluded any other possible cause of renal involvement. Once suspected as the casual relationship between drug and kidney damage, IFN treatment was immediately discontinued. In three cases, we observed a complete and persistent remission of clinical and laboratory abnormalities after IFN withdrawal, while the patient who developed thrombotic microangiopathy, despite IFN withdrawal and complement-inhibitor therapy with eculizumab, showed persistent severe renal failure requiring dialysis.
CONCLUSION This case series highlights the causal relationship between IFN treatment and different types of renal involvement and enables us to delineate several peculiarities of this association.
Collapse
Affiliation(s)
- Iacopo Gianassi
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, Florence 50144, Italy
| | - Marco Allinovi
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, Florence 50144, Italy
| | - Leonardo Caroti
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, Florence 50144, Italy
| | - Lino Calogero Cirami
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, Florence 50144, Italy
| |
Collapse
|
53
|
Davis SE, Khatua AK, Popik W. Nucleosomal dsDNA Stimulates APOL1 Expression in Human Cultured Podocytes by Activating the cGAS/IFI16-STING Signaling Pathway. Sci Rep 2019; 9:15485. [PMID: 31664093 PMCID: PMC6820523 DOI: 10.1038/s41598-019-51998-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/08/2019] [Indexed: 12/31/2022] Open
Abstract
APOL1 alleles G1 and G2 are associated with faster progression to lupus nephritis (LN)-associated end-stage renal disease (LN-ESRD) in African Americans. Increased levels of type I interferons (IFNs) and nucleosome-associated double-stranded DNA (dsDNA) fragments (nsDNA) are the hallmark of this disease. Here, we identify cyclic GMP-AMP synthase (cGAS) and interferon-inducible protein 16 (IFI16) as the major DNA sensors in human immortalized podocytes. We also show that nsDNA triggers the expression of APOL1 and IFNβ via IRF3 activation through the cGAS/IFI16-STING pathway. We demonstrate that maximal APOL1 expression also requires the activation of type I IFN receptor (IFNAR) and STAT1 signaling triggered by IFNβ produced in response to nsDNA, or by exogenous IFNβ. Finally, we show that STAT1 activation is sufficient to upregulate IFI16, subsequently boosting APOL1 expression through a positive feedback mechanism. Collectively, we find that nsDNA-induced APOL1 expression is mediated by both IFNβ-independent and dependent signaling pathways triggered by activation of the cGAS/IFI16-STING pathway. We propose that simultaneous inhibition of STING and the IFNAR-STAT1 pathway may attenuate IFI16 expression, reduce IFI16-cGAS cross-talk, and prevent excessive APOL1 expression in human podocytes in response to nsDNA.
Collapse
Affiliation(s)
- Shamara E Davis
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, Nashville, TN, 37208, USA
| | - Atanu K Khatua
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, Nashville, TN, 37208, USA
| | - Waldemar Popik
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, Nashville, TN, 37208, USA.
- Department of Internal Medicine, 1005 D. B. Todd Blvd, Nashville, TN, 37208, USA.
| |
Collapse
|
54
|
Tanha N, Hansen RB, Yang J, Lange T, Nielsen CT, Helleberg M, Kamper AL, Faurschou M, Jacobsen S. Lymphopenia and neutropenia are associated with subsequent incident proteinuria in Danish patients with systemic lupus erythematosus. Scand J Rheumatol 2019; 49:122-130. [PMID: 31612777 DOI: 10.1080/03009742.2019.1650107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objective: The aim of this study was to investigate whether incident proteinuria in patients with systemic lupus erythematosus (SLE) was preceded by changes in blood lymphocytes and neutrophil counts and/or neutrophil-lymphocyte ratio (NLR).Method: SLE patients with no proteinuria before or at the time of classification were included. Longitudinal data on SLE manifestations, vital status, and SLE-associated medications were collected during clinical visits and chart review. Laboratory data were collected through a nationwide database. Lymphopenia, severe lymphopenia, and neutropenia were defined as values below 0.8 × 109, 0.5 × 109, and 2.0 × 109 cells/L, respectively. High NLR was defined as values above the median. Proteinuria was defined by at least two measurements of elevated urine protein excretion (> 0.5 g/day). Hazard ratios (HRs) were calculated by Cox modelling using time-dependent continuous and binary covariates based on multiple laboratory measurements adjusted for use of immunosuppressants.Results: In total, 260 SLE patients were available for the analysis, of whom 30 (12%) developed incident proteinuria following the diagnosis of SLE. Median follow-up time was 73.5 months. Lymphocyte and neutrophil counts, but not NLR, were associated with incident proteinuria. HRs for incident proteinuria were 2.71 for lymphopenia [95% confidence interval (CI) 1.20-6.11], 4.73 for severe lymphopenia (95% CI 1.93-11.59), and 2.54 for neutropenia (95% CI 1.14-5.65).Conclusion: Lymphopenia and neutropenia predicted the risk of first-time proteinuria independently of immunosuppressants.
Collapse
Affiliation(s)
- N Tanha
- Copenhagen Lupus and Vasculitis Clinic, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - R B Hansen
- Copenhagen Lupus and Vasculitis Clinic, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - J Yang
- Center for Statistical Science, Peking University, Beijing, China
| | - T Lange
- Center for Statistical Science, Peking University, Beijing, China.,Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - C T Nielsen
- Copenhagen Lupus and Vasculitis Clinic, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - M Helleberg
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - A-L Kamper
- Department of Nephrology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - M Faurschou
- Copenhagen Lupus and Vasculitis Clinic, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - S Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
55
|
Adamichou C, Georgakis S, Bertsias G. Cytokine targets in lupus nephritis: Current and future prospects. Clin Immunol 2019; 206:42-52. [DOI: 10.1016/j.clim.2018.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/21/2018] [Accepted: 08/31/2018] [Indexed: 12/13/2022]
|
56
|
Rönnblom L, Leonard D. Interferon pathway in SLE: one key to unlocking the mystery of the disease. Lupus Sci Med 2019; 6:e000270. [PMID: 31497305 PMCID: PMC6703304 DOI: 10.1136/lupus-2018-000270] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/19/2019] [Accepted: 07/30/2019] [Indexed: 12/11/2022]
Abstract
SLE is characterised by an activation of the interferon (IFN) system, which leads to an increased expression of IFN-regulated genes. The reasons behind the IFN signature in SLE are (1) the existence of endogenous IFN inducers, (2) activation of several IFN-producing cell types, (3) production of many different IFNs, (4) a genetic setup promoting IFN production and (5) deficient negative feedback mechanisms. The consequences for the immune system is a continuous stimulation to an immune response, and for the patient a number of different organ manifestations leading to typical symptoms for SLE. In the current review, we will present the existing knowledge of the IFN system and pathway activation in SLE. We will also discuss how this information can contribute to our understanding of both the aetiopathogenesis and some organ manifestations of the disease. We will put forward some issues that are unresolved and should be clarified in order to make a proper stratification of patients with SLE, which seems important when selecting a therapy aiming to downregulate the IFN system.
Collapse
Affiliation(s)
- Lars Rönnblom
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Dag Leonard
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
57
|
Li F, Mao X, Zhuang Q, Zhao Z, Zhang Z, Wu H. Inhibiting 4E-BP1 re-activation represses podocyte cell cycle re-entry and apoptosis induced by adriamycin. Cell Death Dis 2019; 10:241. [PMID: 30858353 PMCID: PMC6411872 DOI: 10.1038/s41419-019-1480-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023]
Abstract
Podocyte loss is one of the determining factors for the progression toward glomerulosclerosis. Podocyte is terminally differentiated and does not typically proliferate following injury and loss. However, recent evidence suggested that during renal injury, podocyte could re-enter the cell cycle, sensitizing the cells to injury and death, but the molecular mechanisms underlying it, as well as the cell fate determination still remained unclear. Here, using NPHS2 Cre; mT/mG transgenic mice and primary podocytes isolated from the mice, we investigated the effect of mammalian target of rapamycin complex 1 (mTORC1)/4E-binding protein 1 (4E-BP1) signaling pathway on cell cycle re-entry and apoptosis of podocyte induced by adriamycin. It was found that podocyte cell cycle re-entry could be induced by adriamycin as early as the 1st week in vivo and the 2nd hour in vitro, accompanied with 4E-BP1 activation and was followed by podocyte loss or apoptosis from the 4th week in vivo or the 4th hour in vitro. Importantly, targeting 4E-BP1 activation by the RNA interference of 4E-BP1 or pharmacologic rapamycin (inhibitor of mTORC1, blocking mTORC1-dependent phosphorylation of its substrate 4E-BP1) treatment was able to inhibit the increases of PCNA, Ki67, and the S-phase fraction of cell cycle in primary podocyte during 2–6 h of adriamycin treatment, and also attenuated the following apoptotic cell death of podocyte detected from the 4th hour, suggesting that 4E-BP1 could be a regulator to manipulate the amount of cell cycle re-entry provided by differentiated podocyte, and thus regulate the degree of podocyte apoptosis, bringing us a new potential podocyte-protective substance that can be used for therapy.
Collapse
Affiliation(s)
- Fang Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xing Mao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiyuan Zhuang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhigang Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Huijuan Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
58
|
Li JS, Li B. Renal Injury Repair: How About the Role of Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:661-670. [PMID: 31399989 DOI: 10.1007/978-981-13-8871-2_32] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Renal failure is one of the most important causes of mortality and morbidity all over the world. Acute kidney injury (AKI) is a major clinical problem that affects up to 5% of all hospitalized patients. Although the kidney has a remarkable capacity for regeneration after acute injury, the mortality among patients with severe AKI remains dismally high, and in clinical practice, most patients cannot be cured completely and suffer from chronic kidney disease (CKD). Recently, the incidence and prevalence of CKD have increased, largely as a result of the enhanced prevalence of diabetes and obesity. The progressive nature of CKD and the ensuing end-stage renal disease (ESRD) place a substantial burden on global healthcare resources. Currently, dialysis and transplantation remain the only treatment options. Finding new therapeutic methods to fight AKI and CKD remains an ongoing quest. Although the human renal histological structure is complex, stem cell therapies have been applied to repair injured kidneys. The curative effects of mesenchymal stem cells (MSCs), hematopoietic stem cells (HSCs), induced pluripotent stem cells (iPSCs), and nephron progenitor cells (NPCs) on renal repair have also been reported by researchers. This review focuses on stem cell therapy and mechanisms for renal injury repair.
Collapse
Affiliation(s)
- Jian-Si Li
- Department of Nephrology, 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Bing Li
- Department of Nephrology, 2nd Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
59
|
Altintas MM, Reiser J. Podocytes: Way to Go. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:226-228. [PMID: 30543788 DOI: 10.1016/j.ajpath.2018.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 11/28/2018] [Indexed: 12/28/2022]
Abstract
This commentary highlights the article by Hara et al that discusses the clinical implications of mitotic catastrophe in podocyte health during diabetic kidney disease.
Collapse
Affiliation(s)
- Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, Illinois.
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
60
|
Qi YY, Zhou XJ, Cheng FJ, Hou P, Ren YL, Wang SX, Zhao MH, Yang L, Martinez J, Zhang H. Increased autophagy is cytoprotective against podocyte injury induced by antibody and interferon-α in lupus nephritis. Ann Rheum Dis 2018; 77:1799-1809. [PMID: 30209031 PMCID: PMC6800572 DOI: 10.1136/annrheumdis-2018-213028] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 07/25/2018] [Accepted: 08/19/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE More recent studies suggested that defects in autophagy contribute to the pathogenesis of SLE, especially in adaptive immunity. Occurrence and progression of lupus nephritis (LN) is the end result of complex interactions between regulation of immune responses and pathological process by renal resident cells, but there is still a lot of missing information for an establishment on the role of autophagy in pathogenesis of LN and as a therapy target. METHODS Systemic and organ-specific aetiologies of autophagy were first evaluated by autophagy protein quantification in tissue homogenates in MRL lpr/lpr lupus prone and female C57BL mice. Analysis of gene expression was also adopted in human blood and urine sediments. Then, some key mediators of the disease, including complement inactivated serum, IgG from patients with LN (IgG-LN) and interferon (IFN)-α were chosen to induce podocyte autophagy. Podocyte injuries including apoptosis, podocin derangement, albumin filtration and wound healing were monitored simultaneously with autophagy steady-state and flux. RESULTS Elevated LC3B in kidney homogenates and increased autophagosomes in podocyte from MRL lpr/lpr were observed. In humans, mRNA levels of some key autophagy genes were increased in blood and urinary sediments, and podocyte autophagosomes were observed in renal biopsies from patients with LN. Complement inactivated serum, IgG-LN and IFN-α could induce podocyte autophagy in a time-dependent and dosage-dependent manner, and by reactive oxygen species production and mTORC1 inhibition, respectively. Autophagy inhibition aggravated podocyte damage whereas its inducer relieved the injury. CONCLUSION Podocyte autophagy is activated in lupus-prone mice and patients with lupus nephritis. Increased autophagy is cytoprotective against antibody and interferon-α induced podocyte injury.
Collapse
Affiliation(s)
- Yuan-Yuan Qi
- Renal Division, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Fa-Juan Cheng
- Renal Division, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Ping Hou
- Renal Division, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Ya-Li Ren
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing, China
| | - Su-Xia Wang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing, China
| | - Ming-Hui Zhao
- Renal Division, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Li Yang
- Renal Division, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, Carolina, USA
| | - Hong Zhang
- Renal Division, Peking University First Hospital; Peking University Institute of Nephrology; Key Laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
61
|
Lee J, Lee J, Kwok S, Baek S, Jang SG, Hong S, Min J, Choi SS, Lee J, Cho M, Park S. JAK
‐1 Inhibition Suppresses Interferon‐Induced
BAFF
Production in Human Salivary Gland. Arthritis Rheumatol 2018; 70:2057-2066. [DOI: 10.1002/art.40589] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/12/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Jaeseon Lee
- The Catholic University of Korea Seoul Republic of Korea
| | - Jennifer Lee
- The Catholic University of Korea Seoul St. Mary's Hospital Seoul Republic of Korea
| | - Seung‐Ki Kwok
- The Catholic University of Korea Seoul St. Mary's Hospital Seoul Republic of Korea
| | - SeungYe Baek
- The Catholic University of Korea Seoul Republic of Korea
| | - Se Gwang Jang
- The Catholic University of Korea Seoul Republic of Korea
| | - Seung‐Min Hong
- The Catholic University of Korea Seoul Republic of Korea
| | - Jae‐Woong Min
- Kangwon National University Chuncheon Republic of Korea
| | - Sun Shim Choi
- Kangwon National University Chuncheon Republic of Korea
| | - Juhyun Lee
- The Catholic University of Korea Seoul Republic of Korea
| | - Mi‐La Cho
- The Catholic University of Korea Seoul Republic of Korea
| | - Sung‐Hwan Park
- The Catholic University of Korea Seoul St. Mary's Hospital Seoul Republic of Korea
| |
Collapse
|
62
|
Voiculescu I, Toda M, Inomata N, Ono T, Li F. Nano and Microsensors for Mammalian Cell Studies. MICROMACHINES 2018; 9:E439. [PMID: 30424372 PMCID: PMC6187600 DOI: 10.3390/mi9090439] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/29/2018] [Accepted: 08/21/2018] [Indexed: 12/20/2022]
Abstract
This review presents several sensors with dimensions at the nano- and micro-scale used for biological applications. Two types of cantilever beams employed as highly sensitive temperature sensors with biological applications will be presented. One type of cantilever beam is fabricated from composite materials and is operated in the deflection mode. In order to achieve the high sensitivity required for detection of heat generated by a single mammalian cell, the cantilever beam temperature sensor presented in this review was microprocessed with a length at the microscale and a thickness in the nanoscale dimension. The second type of cantilever beam presented in this review was operated in the resonant frequency regime. The working principle of the vibrating cantilever beam temperature sensor is based on shifts in resonant frequency in response to temperature variations generated by mammalian cells. Besides the cantilever beam biosensors, two biosensors based on the electric cell-substrate impedance sensing (ECIS) used to monitor mammalian cells attachment and viability will be presented in this review. These ECIS sensors have dimensions at the microscale, with the gold films used for electrodes having thickness at the nanoscale. These micro/nano biosensors and their mammalian cell applications presented in the review demonstrates the diversity of the biosensor technology and applications.
Collapse
Affiliation(s)
- Ioana Voiculescu
- Mechanical Engineering Department, City College of New York, New York, NY 10031, USA.
| | - Masaya Toda
- Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan.
| | - Naoki Inomata
- Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan.
| | - Takahito Ono
- Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan.
| | - Fang Li
- Mechanical Engineering, New York Institute of Technology, New York, NY 11568, USA.
| |
Collapse
|
63
|
Becherucci F, Mazzinghi B, Allinovi M, Angelotti ML, Romagnani P. Regenerating the kidney using human pluripotent stem cells and renal progenitors. Expert Opin Biol Ther 2018; 18:795-806. [PMID: 29939787 DOI: 10.1080/14712598.2018.1492546] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Chronic kidney disease is a major health-care problem worldwide and its cost is becoming no longer affordable. Indeed, restoring damaged renal structures or building a new kidney represents an ambitious and ideal alternative to renal replacement therapy. Streams of research have explored the possible application of pluripotent stem cells (SCs) (embryonic SCs and induced pluripotent SCs) in different strategies aimed at regenerate functioning nephrons and at understanding the mechanisms of kidney regeneration. AREAS COVERED In this review, we will focus on the main potential applications of human pluripotent SCs to kidney regeneration, including those leading to rebuilding new kidneys or part of them (organoids, scaffolds, biological microdevices) as well as those aimed at understanding the pathophysiological mechanisms of renal disease and regenerative processes (modeling of kidney disease, genome editing). Moreover, we will discuss the role of endogenous renal progenitors cells in order to understand and promote kidney regeneration, as an attractive alternative to pluripotent SCs. EXPERT OPINION Opportunities and pitfalls of all these strategies will be underlined, finally leading to the conclusion that a deeper knowledge of the biology of pluripotent SCs is mandatory, in order to allow us to hypothesize their clinical application.
Collapse
Affiliation(s)
- Francesca Becherucci
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy
| | - Benedetta Mazzinghi
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy
| | - Marco Allinovi
- b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Maria Lucia Angelotti
- b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Paola Romagnani
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy.,b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| |
Collapse
|
64
|
Abstract
PURPOSE OF REVIEW Podocytes are critical components of the nephron filtration barrier and are depleted in many kidney injuries and disease states. Terminally differentiated adult podocytes are highly specialized, postmitotic cells, raising the question of whether the body has any ability to regenerate lost podocytes. This timely question has recently been illuminated by a series of innovative studies. Here, we review recent progress on this topic of significant interest and debate. RECENT FINDINGS The innovation of genetic labeling techniques enables fate tracing of individual podocytes, providing the strongest evidence yet that podocytes can be replaced by nearby progenitor cells. In particular, two progenitor pools have recently been identified in multiple studies: parietal epithelial cells and cells of renin lineage. These studies furthermore suggest that podocyte regeneration can be enhanced using ex-vivo or pharmacological interventions. SUMMARY Recent studies indicate that the podocyte compartment is more dynamic than previously believed. Bidirectional exchange with neighboring cellular compartments provides a mechanism for podocyte replacement. Based on these findings, we propose a set of criteria for evaluating podocyte regeneration and suggest that restoration of podocyte number to a subsclerotic threshold be targeted as a potentially achievable clinical goal.
Collapse
|
65
|
Khan SQ, Khan I, Gupta V. CD11b Activity Modulates Pathogenesis of Lupus Nephritis. Front Med (Lausanne) 2018; 5:52. [PMID: 29600248 PMCID: PMC5862812 DOI: 10.3389/fmed.2018.00052] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/13/2018] [Indexed: 01/01/2023] Open
Abstract
Lupus nephritis (LN) is a common complication of systemic lupus erythematosus (SLE) with unclear etiology and limited treatment options. Immune cell infiltration into the kidneys, a hallmark of LN, triggers tissue damage and proteinuria. CD11b, the α-chain of integrin receptor CD11b/CD18 (also known as αMβ2, Mac-1, and CR3), is highly expressed on the surface of innate immune cells, including macrophages and neutrophils. Genetic variants in the human ITGAM gene, which encodes for CD11b, are strongly associated with susceptibility to SLE, LN, and other complications of SLE. CD11b modulates several key biological functions in innate immune cells, including cell adhesion, migration, and phagocytosis. CD11b also modulates other signaling pathways in these cells, such as the Toll-like receptor signaling pathways, that mediate generation of type I interferons, a key proinflammatory cytokine and circulating biomarker in SLE and LN patients. However, how variants in ITGAM gene contribute to disease pathogenesis has not been completely established. Here, we provide an overview of CD11b modulated mechanisms and the functional consequences of the genetic variants that can drive disease pathogenesis. We also present recent insights from studies after pharmacological activation of CD11b. These studies offer novel mechanisms for development of therapeutics for LN, SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Samia Q Khan
- Drug Discovery Center, Department of Internal Medicine, Rush University Medical School, Chicago, IL, United States
| | - Imran Khan
- Drug Discovery Center, Department of Internal Medicine, Rush University Medical School, Chicago, IL, United States
| | - Vineet Gupta
- Drug Discovery Center, Department of Internal Medicine, Rush University Medical School, Chicago, IL, United States
| |
Collapse
|
66
|
Hu W, Niu G, Li H, Gao H, Kang R, Chen X, Lin L. The association between expression of IFIT1 in podocytes of MRL/lpr mice and the renal pathological changes it causes: An animal study. Oncotarget 2018; 7:76464-76470. [PMID: 27823966 PMCID: PMC5363523 DOI: 10.18632/oncotarget.13045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/28/2016] [Indexed: 11/29/2022] Open
Abstract
Renal damage is the major cause of SLE associated mortality, and IFIT1expression was elevated in SLE cases in accordance of previous studies. Therefore, we conducted an animal study to identify the role of IFIT1 expression in renal pathological changes.18 female MRL/lpr mice and same number of female BALB/c mice were enrolled in present study. Quantitative analysis of urine protein, Complement C3 and C4, and anti-ds DNA antibody were conducted. HE and PAS staining and TEM analysis were employed to observe the pathological changes in renal tissue. Significant elevation on urine protein and anti-dsDNA and reduction on Complement C3 and C4 were observed in MRL/lpr mice when comparing the controls in same age. Staining and TEM analysis observed several pathological changes in glomerulus among MRL/lpr mice, including cellular enlargement, basement membrane thickening, and increased cellularcasts. The linear regression analysis found the optical density of IFIT1 was inversely associated with F-actin, Nephrin, and Podocin, but not Synatopodin. In summary, IFIT1 expression is associated with podocytes damage, and capable of suppressing some proteins essential to glomerular filtration.
Collapse
Affiliation(s)
- Weiping Hu
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Guodong Niu
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Hongbo Li
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Hanyuan Gao
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Rudian Kang
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Xiaoqing Chen
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Ling Lin
- Rheumatism Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
67
|
Kim Y, Shim SC. Wolves Trapped in the NETs–The Pathogenesis of Lupus Nephritis. JOURNAL OF RHEUMATIC DISEASES 2018. [DOI: 10.4078/jrd.2018.25.2.81] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Young Kim
- Division of Internal Medicine, Daejeon Veterans Hospital, Daejeon, Korea
| | - Seung Cheol Shim
- Division of Rheumatology, Department of Internal Medicine, Daejeon Rheumatoid and Degenerative Arthritis Center, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
68
|
Chasset F, Arnaud L. Targeting interferons and their pathways in systemic lupus erythematosus. Autoimmun Rev 2017; 17:44-52. [PMID: 29108825 DOI: 10.1016/j.autrev.2017.11.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 09/28/2017] [Indexed: 01/07/2023]
Abstract
Significant advances in the understanding of the molecular basis of innate immunity have led to the identification of interferons (IFNs), particularly IFN-α, as central mediators in the pathogenesis of Systemic Lupus Erythematosus. Therefore, targeting of IFNs and of their downstream pathways has emerged as important developments for novel drug research in SLE. Based on this, several specific interferon blocking strategies using anti-IFN-α antibodies, anti-type I interferon receptor antibodies, Interferon-α-kinoid, or anti-IFN-γ antibodies have all been assessed in recent clinical trials. Alternative strategies targeting the plasmacytoid dendritic cells (pDCs), Toll-Like Receptors (TLRs)-7/9 or their downstream pathways such as the myeloid differentiation primary-response protein 88 (MYD88), spleen tyrosine kinase (Syk), Janus-kinases (JAKs), interleukin-1 receptor-associated kinase 4 (IRAK4), or the Tyrosine Kinase 2 (TYK2) are also investigated actively in SLE, at more preliminary clinical development stages, except for JAK inhibitors which have reached phase 2 studies. In a near future, in-depth and personalized functional characterization of IFN pathways may provide further guidance for the selection of the most relevant therapeutic strategy in SLE, tailored at the patient-level.
Collapse
Affiliation(s)
- François Chasset
- AP-HP, Service de Dermatologie et d'Allergologie, Hôpital Tenon, F-75020, Paris, France
| | - Laurent Arnaud
- Service de rhumatologie, Centre National de Référence des Maladies Autoimmunes et Systémiques Rares, Université de Strasbourg, INSERM UMR-S 1109, F-67000 Strasbourg, France.
| |
Collapse
|
69
|
Sullivan KD, Evans D, Pandey A, Hraha TH, Smith KP, Markham N, Rachubinski AL, Wolter-Warmerdam K, Hickey F, Espinosa JM, Blumenthal T. Trisomy 21 causes changes in the circulating proteome indicative of chronic autoinflammation. Sci Rep 2017; 7:14818. [PMID: 29093484 PMCID: PMC5665944 DOI: 10.1038/s41598-017-13858-3] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/02/2017] [Indexed: 12/11/2022] Open
Abstract
Trisomy 21 (T21) causes Down syndrome (DS), but the mechanisms by which T21 produces the different disease spectrum observed in people with DS are unknown. We recently identified an activated interferon response associated with T21 in human cells of different origins, consistent with overexpression of the four interferon receptors encoded on chromosome 21, and proposed that DS could be understood partially as an interferonopathy. However, the impact of T21 on systemic signaling cascades in living individuals with DS is undefined. To address this knowledge gap, we employed proteomics approaches to analyze blood samples from 263 individuals, 165 of them with DS, leading to the identification of dozens of proteins that are consistently deregulated by T21. Most prominent among these proteins are numerous factors involved in immune control, the complement cascade, and growth factor signaling. Importantly, people with DS display higher levels of many pro-inflammatory cytokines (e.g. IL-6, MCP-1, IL-22, TNF-α) and pronounced complement consumption, resembling changes seen in type I interferonopathies and other autoinflammatory conditions. Therefore, these results are consistent with the hypothesis that increased interferon signaling caused by T21 leads to chronic immune dysregulation, and justify investigations to define the therapeutic value of immune-modulatory strategies in DS.
Collapse
Affiliation(s)
- Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA.,Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Donald Evans
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Ahwan Pandey
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA.,Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | | | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Neil Markham
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Angela L Rachubinski
- JFK Partners/Developmental Pediatrics, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Kristine Wolter-Warmerdam
- Anna and John J. Sie Center for Down Syndrome, Children's Hospital Colorado, Aurora, Colorado, 80045, USA
| | - Francis Hickey
- Anna and John J. Sie Center for Down Syndrome, Children's Hospital Colorado, Aurora, Colorado, 80045, USA
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA. .,Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA. .,Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, 80203, USA.
| | - Thomas Blumenthal
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA. .,Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, 80203, USA. .,Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA.
| |
Collapse
|
70
|
Yu F, Haas M, Glassock R, Zhao MH. Redefining lupus nephritis: clinical implications of pathophysiologic subtypes. Nat Rev Nephrol 2017; 13:483-495. [PMID: 28669995 DOI: 10.1038/nrneph.2017.85] [Citation(s) in RCA: 229] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematosus (SLE) is associated with a broad spectrum of clinical and immunologic manifestations, of which lupus nephritis is the most common cause of morbidity and mortality. The development of nephritis in patients with SLE involves multiple pathogenic pathways including aberrant apoptosis, autoantibody production, immune complex deposition and complement activation. The 2003 International Society of Nephrology/Renal Pathology Society (ISN/RPS) classification system for lupus nephritis was widely accepted with high intraobserver and interobserver concordance to guide therapeutic strategy and provide prognostic information. However, this classification system is not based on the underlying disease pathophysiology. Some additional lesions that contribute to disease presentation, including glomerular crescents, podocyte injury, tubulointerstitial lesions and vascular injury, should be recognized. Although outcomes for patients with lupus nephritis have improved over the past 30 years, treatment of this disease remains challenging and is best approached on the basis of the underlying pathogenesis, which is only partially represented by the various pathological phenotypes defined by the ISN/RPS classification. Here, we discuss the heterogeneous mechanisms involved in the pathogenesis of lupus nephritis and how improved understanding of underlying disease mechanisms might help guide therapeutic strategies.
Collapse
Affiliation(s)
- Feng Yu
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing 100034, P. R. China.,Department of Nephrology, Peking University International Hospital, 1 Zhongguancun Life and Science Street, Changping District, Beijing 102206, P. R. China
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, 90048 California, USA
| | - Richard Glassock
- Department of Medicine, David Geffen School of Medicine at UCLA, 8 Bethany, Laguna Niguel, 92677 California, USA
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Beijing 100034, P. R. China.,Peking-Tsinghua Center for Life Sciences, 5 Summer Palace Street, Haidian District, Beijing 100871, P. R. China
| |
Collapse
|
71
|
Tang H, Lei CT, Ye C, Gao P, Wan C, Chen S, He FF, Wang YM, Su H, Zhang C. MDM2 is implicated in high-glucose-induced podocyte mitotic catastrophe via Notch1 signalling. J Cell Mol Med 2017. [PMID: 28643424 PMCID: PMC5706520 DOI: 10.1111/jcmm.13253] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Podocyte injury and depletion are essential events involved in the pathogenesis of diabetic nephropathy (DN). As a terminally differentiated cell, podocyte is restricted in ‘post‐mitosis’ state and unable to regenerate. Re‐entering mitotic phase will cause podocyte disastrous death which is defined as mitotic catastrophe (MC). Murine double minute 2 (MDM2), a cell cycle regulator, is widely expressed in renal resident cells including podocytes. Here, we explore whether MDM2 is involved in podocyte MC during hyperglycaemia. We found aberrant mitotic podocytes with multi‐nucleation in DN patients. In vitro, cultured podocytes treated by high glucose (HG) also showed an up‐regulation of mitotic markers and abnormal mitotic status, accompanied by elevated expression of MDM2. HG exposure forced podocytes to enter into S phase and bypass G2/M checkpoint with enhanced expression of Ki67, cyclin B1, Aurora B and p‐H3. Genetic deletion of MDM2 partly reversed HG‐induced mitotic phase re‐entering of podocytes. Moreover, HG‐induced podocyte injury was alleviated by MDM2 knocking down but not by nutlin‐3a, an inhibitor of MDM2‐p53 interaction. Interestingly, knocking down MDM2 or MDM2 overexpression showed inhibition or activation of Notch1 signalling, respectively. In addition, genetic silencing of Notch1 prevented HG‐mediated podocyte MC. In conclusion, high glucose up‐regulates MDM2 expression and leads to podocyte MC. Notch1 signalling is an essential downstream pathway of MDM2 in mediating HG‐induced MC in podocytes.
Collapse
Affiliation(s)
- Hui Tang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Tao Lei
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Ye
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Gao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Wan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Chen
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang-Fang He
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Mei Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
72
|
Elmaghrabi A, Brown E, Khin E, Hassler J, Hendricks AR. Tubuloreticular Inclusions in the Absence of Systemic Lupus Erythematosus and HIV Infection: A Report of Three Pediatric Cases. Case Rep Nephrol Dial 2017; 7:91-101. [PMID: 28868299 PMCID: PMC5567081 DOI: 10.1159/000477661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/21/2017] [Indexed: 11/19/2022] Open
Abstract
Tubuloreticular inclusions (TRIs) are subcellular structures located within the cisternae of endoplasmic reticulum. Formation of TRIs has been linked to the exposure of excess interferon (IFN), either from endogenous or exogenous sources. In renal disease, TRIs have been most commonly associated with systemic lupus erythematosus (SLE), and human immunodeficiency virus-associated nephropathy (HIVAN). Case reports of patients with renal biopsies showing TRIs without underlying SLE or HIV are infrequent in adults, and to our knowledge none have been reported in children. We report 3 pediatric cases in which the renal biopsy showed TRIs on electron microscopy without underlying SLE or HIV infection. The first patient presented at 2 years of age with nephrotic syndrome and renal failure. His renal biopsy revealed focal segmental glomerulosclerosis and TRIs. The second patient presented at 6 months of age with infantile nephrotic syndrome, and his renal biopsy revealed membranous glomerulopathy and TRIs. The last patient presented at 4 years of age with acute kidney injury of unclear etiology leading to chronic kidney disease. Her biopsy revealed acute and chronic tubulointerstitial nephritis with TRIs. Despite extensive evaluation in all 3 patients, including testing for HIV infection and SLE, we could not identify an underlying etiology to explain the presence of TRIs. In conclusion, renal biopsy with TRIs in the absence of underling SLE and HIV remains obscure. We propose a possible role for excess IFN triggered by an abnormal immune response to common viral infections in the formation of TRIs and renal injury.
Collapse
Affiliation(s)
- Ayah Elmaghrabi
- Children's Medical Center, Dallas, Texas, USA.,UT Southwestern Medical Center, Dallas, Texas, USA
| | - Elizabeth Brown
- Children's Medical Center, Dallas, Texas, USA.,UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ei Khin
- Children's Medical Center, Dallas, Texas, USA.,UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jared Hassler
- Children's Medical Center, Dallas, Texas, USA.,UT Southwestern Medical Center, Dallas, Texas, USA
| | - Allen R Hendricks
- Children's Medical Center, Dallas, Texas, USA.,UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
73
|
Abstract
In normal glomeruli, parietal epithelial cells (PECs) line the inside of Bowman's capsule and form an inconspicuous sheet of flat epithelial cells in continuity with the proximal tubular epithelial cells (PTECs) at the urinary pole and with the podocytes at the vascular pole. PECs, PTECs and podocytes have a common mesenchymal origin and are the result of divergent differentiation during embryogenesis. Podocytes and PTECs are highly differentiated cells with well-established functions pertaining to the maintenance of the filtration barrier and transport, respectively. For PECs, no specific function other than a structural one has been known until recently. Possible important functions for PECs in the fate of the glomerulus in glomerular disease have now become apparent: (1) PECs may be involved in the replacement of lost podocytes; (2) PECs form the basis of extracapillary proliferative lesions and subsequent sclerosis in glomerular disease. In addition to the acknowledgement that PECs are crucial in glomerular disease, knowledge has been gained regarding the molecular processes driving the phenotypic changes and behavior of PECs. Understanding these molecular processes is important for the development of specific therapeutic approaches aimed at either stimulation of the regenerative function of PECs or inhibition of the pro-sclerotic action of PECs. In this review, we discuss recent advances pertaining to the role of PECs in glomerular regeneration and disease and address the major molecular processes involved.
Collapse
|
74
|
Lan X, Wen H, Cheng K, Plagov A, Marashi Shoshtari SS, Malhotra A, Singhal PC. Hedgehog pathway plays a vital role in HIV-induced epithelial-mesenchymal transition of podocyte. Exp Cell Res 2017; 352:193-201. [PMID: 28159470 DOI: 10.1016/j.yexcr.2017.01.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/25/2017] [Accepted: 01/29/2017] [Indexed: 11/28/2022]
Abstract
HIV-associated nephropathy (HIVAN) is characterized by heavy proteinuria, rapidly progressive renal failure, and distinct morphological features in the kidney. HIV-induced epithelial-mesenchymal transition (EMT) is critically important for the progression of kidney injury. In this study, we tested the role of hedgehog pathway in the HIV-induced EMT and fibrosis of kidney. We used the Tg26 mice, the abundantly used HIVAN mouse model, to investigate the activation of hedgehog pathway by HIV. Western blotting and real time PCR results showed that renal tissue expression of hedgehog pathway related molecules, including hedgehog homologous (Shh, Ihh, Dhh), PTCH, and Gli1, were increased in HIVAN (Tg26) mice; while immunofluorescent staining displayed localization PTCH expression in podocytes. For in vitro studies, we used recombinant sonic hedgehog (Shh) and HIV for their expression by podocytes. Both the methods activated the hedgehog pathway, enhanced the expression of EMT markers, and decreased impermeability. Overexpression of Gli1 by human podocytes also augmented their expression of EMT markers. On the other hand, the blockade of hedgehog pathway with Gant 58, a specific blocker for Gli1-induced transcription, dramatically decreased HIV-induced podocyte EMT and permeability. These results indicate that hedgehog pathway plays an important role in HIV-induced podocyte injury. The present study provides mechanistical insight into a new target for therapeutic strategy.
Collapse
Affiliation(s)
- Xiqian Lan
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra Northwell Medical School, NY, USA.
| | - Hongxiu Wen
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra Northwell Medical School, NY, USA
| | - Kang Cheng
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra Northwell Medical School, NY, USA
| | - Andrei Plagov
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra Northwell Medical School, NY, USA
| | | | - Ashwani Malhotra
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra Northwell Medical School, NY, USA
| | - Pravin C Singhal
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra Northwell Medical School, NY, USA.
| |
Collapse
|
75
|
Abstract
Finding better treatments for lupus nephritis requires an understanding of the pathogenesis of the causative systemic disease, how this leads to kidney disease, and how lupus nephritis progresses to end-stage kidney disease. Here, we provide a brief conceptual overview on the related pathomechanisms. As a main focus we discuss in detail the roles of neutrophils, dendritic cells, Toll-like receptors, and interferon-α in the pathogenesis of lupus nephritis by separately reviewing their roles in extrarenal systemic autoimmunity and in intrarenal inflammation and immunopathology.
Collapse
|
76
|
Mazzinghi B, Romagnani P, Lazzeri E. Biologic modulation in renal regeneration. Expert Opin Biol Ther 2016; 16:1403-1415. [DOI: 10.1080/14712598.2016.1219336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
77
|
Ma R, Jiang W, Li Z, Sun Y, Wei Z. Intrarenal macrophage infiltration induced by T cells is associated with podocyte injury in lupus nephritis patients. Lupus 2016; 25:1577-1586. [PMID: 27147620 DOI: 10.1177/0961203316646861] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/04/2016] [Indexed: 11/16/2022]
Abstract
Proteinuria is the hallmark of clinical manifestation of disease activity in lupus nephritis (LN) patients, which arises from direct or indirect podocyte injury. This study is to explore the relationship between intrarenal T cell infiltration and podocyte injury in lupus nephritis (LN), and to understand the potential mechanisms of podocyte injury induced by intrarenal T cells. Sixty renal biopsies from patients diagnosed with LN were included in the present study. Histological changes in LN patients were detected by light and electron microscopy. Podocyte-specific nephrin expression in renal tissues was detected by immunofluorescence. Infiltration of T cells (CD3+ cells), infiltration of macrophages (CD68+ cells) and the expression of osteopontin (OPN) in renal tissues were examined by immunohistochemical staining. Pearson or Spearman’s tests were used to perform correlation analysis. Morphologic lesions of podocytes were more severe in LN patients than in normal control subjects. Compared with normal control subjects, nephrin expression was significantly decreased in LN patients. The expression level of nephrin was significantly lower in active LN patients than in the inactive group of patients ( P < 0.05). Compared with normal control subjects, the number of infiltrated intrarenal T cells and macrophages was significantly increased in LN patients. T cells were mainly distributed in renal interstitium, with very few being in glomeruli, while macrophages were mainly located in glomeruli. The number of intrarenal infiltrated T cells and macrophages in active LN patients was more than that in the inactive group ( P < 0.05). Compared with normal control subjects, OPN expression in LN patients was increased significantly. The expression level of OPN in active LN patients was significantly higher than that in the inactive group ( P < 0.05). Podocyte-specific nephrin was negatively correlated with 24-hour proteinuria, intrarenal T cells infiltration and intrarenal OPN expression in LN patients ( P < 0.001). Intrarenal macrophages had significantly positive correlation with intrarenal OPN expression ( P < 0.001). The present study provides possible links between intrarenal T cells, OPN, macrophages with reduced podocyte-nephrin and podocytopathy in systemic lupus erythematosus. In addition, infiltration of macrophages in glomeruli induced by OPN that is induced by T cells may be a crucial mechanism for podocyte injury.
Collapse
Affiliation(s)
- R Ma
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - W Jiang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Z Li
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Y Sun
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Z Wei
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, PR China
| |
Collapse
|
78
|
Hagen M, Pfister E, Kosel A, Shankland S, Pippin J, Amann K, Daniel C. Cell cycle re-entry sensitizes podocytes to injury induced death. Cell Cycle 2016; 15:1929-37. [PMID: 27232327 DOI: 10.1080/15384101.2016.1191710] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Podocytes are terminally differentiated renal cells, lacking the ability to regenerate by proliferation. However, during renal injury, podocytes re-enter into the cell cycle but fail to divide. Earlier studies suggested that re-entry into cell cycle results in loss of podocytes, but a direct evidence for this is lacking. Therefore, we established an in vitro model to test the consequences of re-entry into the cell cycle on podocyte survival. A mouse immortalized podocyte cell line was differentiated to non-permissive podocytes and stimulated with e.g. growth factors. Stimulated cells were analyzed for mRNA-expression or stained for cell cycle analysis using flow cytometry and immunocytofluorescence microscopy. After stimulation to re-entry into cell cycle, podocytes were stressed with puromycin aminonucleoside (PAN) and analyzed for survival. During permissive stage more than 40% of immortalized podocytes were in the S-phase. In contrast, S-phase in non-permissive differentiated podocytes was reduced to 5%. Treatment with b-FGF dose dependently induced re-entry into cell cycle increasing the number of podocytes in the S-phase to 10.7% at an optimal bFGF dosage of 10 ng/ml. Forty eight hours after stimulation with bFGF the number of bi-nucleated podocytes significantly increased. A secondary injury stimulus significantly reduced podocyte survival preferentially in bi-nucleated podocytes In conclusion, stimulation of podocytes using bFGF was able to induce re-entry of podocytes into the cell cycle and to sensitize the cells for cell death by secondary injuries. Therefore, this model is appropriate for testing new podocyte protective substances that can be used for therapy.
Collapse
Affiliation(s)
- Manuel Hagen
- a Department of Nephropathology , Friedrich-Alexander University (FAU) Erlangen-Nürnberg , Erlangen , Germany
| | - Eva Pfister
- a Department of Nephropathology , Friedrich-Alexander University (FAU) Erlangen-Nürnberg , Erlangen , Germany
| | - Andrea Kosel
- a Department of Nephropathology , Friedrich-Alexander University (FAU) Erlangen-Nürnberg , Erlangen , Germany
| | - Stuart Shankland
- b Department of Nephrology , University of Washington , Seattle , WA , USA
| | - Jeffrey Pippin
- b Department of Nephrology , University of Washington , Seattle , WA , USA
| | - Kerstin Amann
- a Department of Nephropathology , Friedrich-Alexander University (FAU) Erlangen-Nürnberg , Erlangen , Germany
| | - Christoph Daniel
- a Department of Nephropathology , Friedrich-Alexander University (FAU) Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
79
|
Links between coagulation, inflammation, regeneration, and fibrosis in kidney pathology. J Transl Med 2016; 96:378-90. [PMID: 26752746 DOI: 10.1038/labinvest.2015.164] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/22/2015] [Indexed: 12/14/2022] Open
Abstract
Acute kidney injury (AKI) involves nephron injury leading to irreversible nephron loss, ie, chronic kidney disease (CKD). Both AKI and CKD are associated with distinct histological patterns of tissue injury, but kidney atrophy in CKD involves tissue remodeling with interstitial inflammation and scarring. No doubt, nephron atrophy, inflammation, fibrosis, and renal dysfunction are associated with each other, but their hierarchical relationships remain speculative. To better understand the pathophysiology, we provide an overview of the fundamental danger response programs that assure host survival upon traumatic injury from as early as the first multicellular organisms, ie, bleeding control by coagulation, infection control by inflammation, epithelial barrier restoration by re-epithelialization, and tissue stabilization by mesenchymal repair. Although these processes assure survival in the majority of the populations, their dysregulation causes kidney disease in a minority. We discuss how, in genetically heterogeneous population, genetic variants shift balances and modulate danger responses toward kidney disease. We further discuss how classic kidney disease entities develop from an insufficient or overshooting activation of these danger response programs. Finally, we discuss molecular pathways linking, for example, inflammation and regeneration or inflammation and fibrosis. Understanding the causative and hierarchical relationships and the molecular links between the danger response programs should help to identify molecular targets to modulate kidney injury and to improve outcomes for kidney disease patients.
Collapse
|
80
|
Abstract
Despite marked improvements in the survival of patients with severe lupus nephritis over the past 50 years, the rate of complete clinical remission after immune suppression therapy is <50% and renal impairment still occurs in 40% of affected patients. An appreciation of the factors that lead to the development of chronic kidney disease following acute or subacute renal injury in patients with systemic lupus erythematosus is beginning to emerge. Processes that contribute to end-stage renal injury include continuing inflammation, activation of intrinsic renal cells, cell stress and hypoxia, metabolic abnormalities, aberrant tissue repair and tissue fibrosis. A deeper understanding of these processes is leading to the development of novel or adjunctive therapies that could protect the kidney from the secondary non-immune consequences of acute injury. Approaches based on a molecular-proteomic-lipidomic classification of disease should yield new information about the functional basis of disease heterogeneity so that the most effective and least toxic treatment regimens can be formulated for individual patients.
Collapse
|
81
|
Kumar Vr S, Darisipudi MN, Steiger S, Devarapu SK, Tato M, Kukarni OP, Mulay SR, Thomasova D, Popper B, Demleitner J, Zuchtriegel G, Reichel C, Cohen CD, Lindenmeyer MT, Liapis H, Moll S, Reid E, Stitt AW, Schott B, Gruner S, Haap W, Ebeling M, Hartmann G, Anders HJ. Cathepsin S Cleavage of Protease-Activated Receptor-2 on Endothelial Cells Promotes Microvascular Diabetes Complications. J Am Soc Nephrol 2015; 27:1635-49. [PMID: 26567242 DOI: 10.1681/asn.2015020208] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 08/25/2015] [Indexed: 01/13/2023] Open
Abstract
Endothelial dysfunction is a central pathomechanism in diabetes-associated complications. We hypothesized a pathogenic role in this dysfunction of cathepsin S (Cat-S), a cysteine protease that degrades elastic fibers and activates the protease-activated receptor-2 (PAR2) on endothelial cells. We found that injection of mice with recombinant Cat-S induced albuminuria and glomerular endothelial cell injury in a PAR2-dependent manner. In vivo microscopy confirmed a role for intrinsic Cat-S/PAR2 in ischemia-induced microvascular permeability. In vitro transcriptome analysis and experiments using siRNA or specific Cat-S and PAR2 antagonists revealed that Cat-S specifically impaired the integrity and barrier function of glomerular endothelial cells selectively through PAR2. In human and mouse type 2 diabetic nephropathy, only CD68(+) intrarenal monocytes expressed Cat-S mRNA, whereas Cat-S protein was present along endothelial cells and inside proximal tubular epithelial cells also. In contrast, the cysteine protease inhibitor cystatin C was expressed only in tubules. Delayed treatment of type 2 diabetic db/db mice with Cat-S or PAR2 inhibitors attenuated albuminuria and glomerulosclerosis (indicators of diabetic nephropathy) and attenuated albumin leakage into the retina and other structural markers of diabetic retinopathy. These data identify Cat-S as a monocyte/macrophage-derived circulating PAR2 agonist and mediator of endothelial dysfunction-related microvascular diabetes complications. Thus, Cat-S or PAR2 inhibition might be a novel strategy to prevent microvascular disease in diabetes and other diseases.
Collapse
Affiliation(s)
- Santhosh Kumar Vr
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Murthy N Darisipudi
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Stefanie Steiger
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Satish Kumar Devarapu
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Maia Tato
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Onkar P Kukarni
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Shrikant R Mulay
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Dana Thomasova
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Bastian Popper
- Department of Anatomy and Cell Biology, Ludwig-Maximilians Universität, Munich, Germany
| | | | - Gabriele Zuchtriegel
- Walter Brendel Centre of Experimental Medicine, and Department of Otorhinolaryngology, Head and Neck Surgery, University of Munich, Munich, Germany
| | - Christoph Reichel
- Walter Brendel Centre of Experimental Medicine, and Department of Otorhinolaryngology, Head and Neck Surgery, University of Munich, Munich, Germany
| | - Clemens D Cohen
- Division of Nephrology, Krankenhaus Harlaching, Munich, Germany; Division of Nephrology and Institute of Physiology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | - Helen Liapis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Solange Moll
- Institute of Clinical Pathology, University Hospital Geneva, Geneva, Switzerland
| | - Emma Reid
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Ireland; and
| | - Alan W Stitt
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Ireland; and
| | - Brigitte Schott
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Sabine Gruner
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Wolfgang Haap
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Martin Ebeling
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Guido Hartmann
- Cardiovascular and Metabolism, Pharma Research and Early Development, Hoffmann La Roche, Basel, Switzerland
| | - Hans-Joachim Anders
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, Munich, Germany;
| |
Collapse
|
82
|
Abstract
INTRODUCTION Kidney diseases are a global public health problem whose incidence is rapidly growing due to a global rise in the aged population and the increasing prevalence of cardiovascular disease, hypertension and diabetes. With the emergence of stem cells as potential therapeutic agents, attempts in using them to significantly reduce the burden of these diseases have increased. AREAS COVERED Several types of stem cells have been proven to be likely candidates for treating kidney diseases. We discuss in detail the potential use of mesenchymal stem cells in preclinical and clinical works, with additional populations that have been studied briefly described. Moreover, we discuss current knowledge on endogenous kidney regeneration ability and on the possibility to modulate it using chemical and biological agents. EXPERT OPINION Stem cell therapy is a promising new treatment for kidney disease documented in many animal studies. Mesenchymal stem cells have emerged as a promising cell type, but their efficacy in clinical trials is still controversial. Identification of progenitor cells in the adult kidney is another step forward in regenerative medicine, suggesting the repair potential of the adult kidney and the possible modulation of renal progenitors in situ using pharmacological approaches.
Collapse
Affiliation(s)
- Elena Lazzeri
- a University of Florence; Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE) , Florence, Italy +390552758342 ; ;
| | - Paola Romagnani
- a University of Florence; Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE) , Florence, Italy +390552758342 ; ;
| | - Laura Lasagni
- a University of Florence; Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE) , Florence, Italy +390552758342 ; ;
| |
Collapse
|
83
|
A Plasmacytoid Dendritic Cells-Type I Interferon Axis Is Critically Implicated in the Pathogenesis of Systemic Lupus Erythematosus. Int J Mol Sci 2015; 16:14158-70. [PMID: 26110387 PMCID: PMC4490545 DOI: 10.3390/ijms160614158] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/01/2015] [Accepted: 06/16/2015] [Indexed: 01/12/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease that is characterized by the generation of immune responses to various nuclear components. Impaired clearance of apoptotic cells and loss of tolerance to self-antigens are involved both in the initiation and in the propagation of the disease. Dendritic cells (DCs) are key factors in the balance between autoimmunity and tolerance and play a role linking innate and adaptive immunity. DCs, particularly plasmacytoid DCs (pDCs), are the main source of type I interferon (IFN) cytokines, which contribute to the immunopathogenesis of SLE. There is accumulating evidence that pDCs and type I IFN cytokines take the leading part in the development of SLE. In this review, we discuss recent data regarding the role of pDCs and type I IFN cytokines in the pathogenesis of SLE and the potential for employing therapies targeting against aberrant regulation of the pDC-type I IFN axis for treating SLE.
Collapse
|
84
|
Advances in understanding the role of type I interferons in systemic lupus erythematosus. Curr Opin Rheumatol 2015; 26:467-74. [PMID: 25010440 DOI: 10.1097/bor.0000000000000087] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Advances in understanding the genetic and molecular basis of innate immune system activation and function have supported the hypothesis that type I interferons (IFN-I), the essential mediators of antiviral host defense, are central contributors to the pathogenesis of systemic lupus erythematosus (SLE). This review addresses the recent data that support the rationale for therapeutic targeting of the IFN-I pathway in SLE. RECENT FINDINGS New insights into the mechanisms of cell-intrinsic innate immune system activation, driven by endogenous virus-like nucleic acids and potentially modified by environmental stressors, provide a model for the induction of IFN-I that may precede the clinically apparent autoimmunity in patients with lupus. Further amplification of IFN-α production, induced by nucleic-acid-containing immune complexes that activate endosomal Toll-like receptors, augments and sustains immune system activation, autoimmunity and tissue damage. SUMMARY As demonstrated in the murine studies of persistent virus infection accompanied by sustained production of IFN-I, blockade of the IFN-I pathway may reverse the immune dysregulation and tissue damage that are the essential features of the immunopathogenesis of SLE. Recent research progress has identified numerous therapeutic targets, and specific candidate therapeutics relevant to the IFN-I pathway are under investigation.
Collapse
|
85
|
Quintana LF, Campistol JM. Interferón ß, microangiopatía trombótica y síndrome nefrótico. Med Clin (Barc) 2015; 144:65-6. [DOI: 10.1016/j.medcli.2014.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 09/25/2014] [Indexed: 10/24/2022]
|
86
|
Zhang D, Wei G, Li P, Zhou X, Zhang Y. Urine-derived stem cells: A novel and versatile progenitor source for cell-based therapy and regenerative medicine. Genes Dis 2014; 1:8-17. [PMID: 25411659 PMCID: PMC4234168 DOI: 10.1016/j.gendis.2014.07.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Engineered functional organs or tissues, created with autologous somatic cells and seeded on biodegradable or hydrogel scaffolds, have been developed for use in individuals with tissue damage suffered from congenital disorders, infection, irradiation, or cancer. However, in those patients, abnormal cells obtained by biopsy from the compromised tissue could potentially contaminate the engineered tissues. Thus, an alternative cell source for construction of the neo-organ or functional recovery of the injured or diseased tissues would be useful. Recently, we have found stem cells existing in the urine. These cells are highly expandable, and have self-renewal capacity, paracrine properties, and multi-differentiation potential. As a novel cell source, urine-derived stem cells (USCs) provide advantages for cell therapy and tissue engineering applications in regeneration of various tissues, particularly in the genitourinary tract, because they originate from the urinary tract system. Importantly, USCs can be obtained via a non-invasive, simple, and low-cost approach and induced with high efficiency to differentiate into three dermal cell lineages.
Collapse
Affiliation(s)
- Deying Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China ; Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Peng Li
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA ; Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaobo Zhou
- Center for Bioinformatics and Systems Biology, Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
87
|
Abstract
Investigations of patients with systemic lupus erythematosus have applied insights from studies of the innate immune response to define IFN-I, with IFN-α as the dominant mediator, as central to the pathogenesis of this prototype systemic autoimmune disease. Genetic association data identify regulators of nucleic acid degradation and components of TLR-independent, endosomal TLR-dependent, and IFN-I-signaling pathways as contributors to lupus disease susceptibility. Together with a gene expression signature characterized by IFN-I-induced gene transcripts in lupus blood and tissue, those data support the conclusion that many of the immunologic and pathologic features of this disease are a consequence of a persistent self-directed immune reaction driven by IFN-I and mimicking a sustained antivirus response. This expanding knowledge of the role of IFN-I and the innate immune response suggests candidate therapeutic targets that are being tested in lupus patients.
Collapse
Affiliation(s)
- Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, NY 10021; and Rheumatology Division, Department of Medicine, NewYork-Presbyterian/Weill Cornell Medical Center, New York, NY 10065
| |
Collapse
|
88
|
Abkhezr M, Dryer SE. Angiotensin II and canonical transient receptor potential-6 activation stimulate release of a signal transducer and activator of transcription 3-activating factor from mouse podocytes. Mol Pharmacol 2014; 86:150-8. [PMID: 24850910 DOI: 10.1124/mol.114.092536] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Previous studies have shown that the transcription factor signal transducer and activator of transcription-3 (STAT3) in podocytes plays an important role in progression of HIV nephropathy and in collapsing forms of glomerulonephritis. Here, we have observed that application of 100 nM angiotensin II (Ang II) to cultured podocytes for 6-24 hours causes a marked increase in the phosphorylation of STAT3 on tyrosine Y705 but has no effect on phosphorylation at serine S727. By contrast, Ang II treatment of short periods (20-60 minutes) caused a small but consistent suppression of tyrosine phosphylation of STAT3. A similar biphasic effect was seen after treatment with the diacylglycerol analog 1-oleoyl-2-acetyl-sn-glycerol (OAG), an agent that causes activation of Ca(2+)-permeable canonical transient receptor potential-6 (TRPC6) channels in podocytes. The stimulatory effects of Ang II on STAT3 phosphorylation were abolished by small-interfering RNA knockdown of TRPC6 and also by inhibitors of the Ca(2+)-dependent downstream enzymes calcineurin and Ca(2+)-calmodulin-dependent protein kinase II. The stimulatory effects of Ang II appear to be mediated by secretion and accumulation of an unknown factor into the surrounding medium, as they are no longer detected when medium is replaced every 2 hours even if Ang II is continuously present. By contrast, the inhibitory effect of Ang II on STAT3 phosphorylation persists with frequent medium changes. Experiments with neutralizing and inhibitory antibodies suggest that the STAT3 stimulatory factor secreted from podocytes is not interleukin-6, but also suggest that this factor exerts its actions through a receptor system that requires glycoprotein 130.
Collapse
Affiliation(s)
- Mousa Abkhezr
- Department of Biology and Biochemistry, University of Houston (M.A., S.E.D.), and Division of Nephrology, Baylor College of Medicine (S.E.D.), Houston, Texas
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston (M.A., S.E.D.), and Division of Nephrology, Baylor College of Medicine (S.E.D.), Houston, Texas
| |
Collapse
|
89
|
Anders HJ, Schaefer L. Beyond tissue injury-damage-associated molecular patterns, toll-like receptors, and inflammasomes also drive regeneration and fibrosis. J Am Soc Nephrol 2014; 25:1387-400. [PMID: 24762401 PMCID: PMC4073442 DOI: 10.1681/asn.2014010117] [Citation(s) in RCA: 223] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tissue injury initiates an inflammatory response through the actions of immunostimulatory molecules referred to as damage-associated molecular patterns (DAMPs). DAMPs encompass a group of heterogenous molecules, including intracellular molecules released during cell necrosis and molecules involved in extracellular matrix remodeling such as hyaluronan, biglycan, and fibronectin. Kidney-specific DAMPs include crystals and uromodulin released by renal tubular damage. DAMPs trigger innate immunity by activating Toll-like receptors, purinergic receptors, or the NLRP3 inflammasome. However, recent evidence revealed that DAMPs also trigger re-epithelialization upon kidney injury and contribute to epithelial-mesenchymal transition and, potentially, to myofibroblast differentiation and proliferation. Thus, these discoveries suggest that DAMPs drive not only immune injury but also kidney regeneration and renal scarring. Here, we review the data from these studies and discuss the increasingly complex connection between DAMPs and kidney diseases.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Nephrological Center, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany; and
| | - Liliana Schaefer
- Pharmazentrum Frankfurt, Institute of General Pharmacology and Toxicology, Goethe-University of Frankfurt/Main, Frankfurt/Main, Germany
| |
Collapse
|
90
|
Abstract
The very limited ability of adult podocytes to proliferate in vivo is clinically significant because podocytes form a vascular barrier that is functionally critical to the nephron, podocyte hypoplasia is a characteristic of disease, and inadequate regeneration of podocytes is a major cause of persistent podocyte hypoplasia. Excessive podocyte loss or inadequate replacement leads to glomerulosclerosis in many progressive kidney diseases. Thus, restoration of podocyte cell density almost certainly is reliant on regeneration by podocyte progenitors. However, such putative progenitors have remained elusive until recently. In this review, we describe the developmental processes leading to podocyte and parietal epithelial cell (PEC) formation during glomerulogenesis. We compare evidence that in normal human kidneys PECs expressing progenitor markers CD133 and CD24 can differentiate into podocytes in vitro and in vivo, with evidence from animal models suggesting a more limited role of the PEC's capacity to serve as a podocyte progenitor in adults. We highlight tantalizing new evidence that specialized vascular wall cells of afferent arterioles, including those that produce renin in healthy kidney, provide a novel local progenitor source of new PECs and podocytes in response to podocyte hypoplasia in the adult, and draw comparisons with glomerulogenesis.
Collapse
Affiliation(s)
- Stuart J Shankland
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA.
| | - Jeffrey W Pippin
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | | |
Collapse
|
91
|
Anders HJ. Immune system modulation of kidney regeneration--mechanisms and implications. Nat Rev Nephrol 2014; 10:347-58. [PMID: 24776845 DOI: 10.1038/nrneph.2014.68] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The immune system is an important guardian of tissue homeostasis. In response to injury, resident and infiltrating immune cells orchestrate all phases of danger control, resolution of inflammation and tissue regeneration or scar formation. As mammalian postnatal kidneys are not capable of de novo nephrogenesis, recovery is limited to the regeneration or repair of existing nephrons. The regenerative capacity of the nephron varies between compartments; the epithelial cells of the tubule regenerate more efficiently than the structurally highly organized podocytes. Cells of the surrounding environment modulate nephron regeneration by secreting paracrine mediators. This Review discusses immune mediators and pathways that regulate the intrinsic regenerative capacity of the nephron. Eliminating injurious triggers, modulating renal inflammation and specifically enhancing the regenerative capacity of nephrons might be a promising strategy to improve long-term outcomes in patients with acute kidney injury and/or chronic kidney disease.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München-Innenstadt, Ziemssenstrasse 1, 80336 Munich, Germany
| |
Collapse
|
92
|
Yuste C, Rapalai M, Pritchard BA, Jones TJ, Tucker B, Ramakrishna SB. Nephrotic-range proteinuria on interferon-β treatment: immune-induced glomerulonephritis or other pathway? Clin Kidney J 2014; 7:190-3. [PMID: 25852870 PMCID: PMC4377790 DOI: 10.1093/ckj/sfu016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 02/13/2014] [Indexed: 12/22/2022] Open
Abstract
We present a case report of a 37-year-old woman with multiple sclerosis (MS) who developed nephrotic-range proteinuria secondary to membrano proliferative glomerulonephritis (MPGN)-like disease with mesangial C3 deposition without evidence of immune-complex deposition in the context of long-term interferon-β (IFN-β) therapy. The complete remission of proteinuria following cessation of IFN-β, strongly suggests causality. To our knowledge, this is the second case report of MPGN associated with IFN-β use. This being the case, the negative immune screen, normal inflammatory markers and the absence of immune complex deposits would imply a different pathway to that previously suggested.
Collapse
Affiliation(s)
- C Yuste
- Shrewsbury and Telford NHS Trust , Shorpshire, UK
| | - M Rapalai
- Shrewsbury and Telford NHS Trust , Shorpshire, UK
| | | | - T J Jones
- Shrewsbury and Telford NHS Trust , Shorpshire, UK
| | - B Tucker
- King's College Hospital , London, UK
| | | |
Collapse
|
93
|
Shankland SJ, Smeets B, Pippin JW, Moeller MJ. The emergence of the glomerular parietal epithelial cell. Nat Rev Nephrol 2014; 10:158-73. [PMID: 24468766 DOI: 10.1038/nrneph.2014.1] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glomerular diseases are the leading causes of chronic and end-stage kidney disease. In the 1980s and 1990s, attention was focused on the biology and role of glomerular endothelial and mesangial cells. For the past two decades, seminal discoveries have been made in podocyte biology in health and disease. More recently, the glomerular parietal epithelial cell (PEC)-the fourth resident glomerular cell type-has been under active study, leading to a better understanding and definition of how these cells behave normally, and their potential roles in glomerular disease. Accordingly, this Review will focus on our current knowledge of PECs, in both health and disease. We discuss model systems to study PECs, how PECs might contribute to glomerulosclerosis, crescent and pseudocrescent formation and how PECs handle filtered albumin. These events have consequences on PEC structure and function, and PECs have potential roles as stem or progenitor cells for podocytes in glomerular regeneration, which will also be described.
Collapse
Affiliation(s)
- Stuart J Shankland
- Division of Nephrology, University of Washington, 1959 North East Pacific Avenue, Box 356521, Room BB1269, Seattle, WA 98195-6521, USA
| | - Bart Smeets
- Nephrology and Clinical Immunology, University Hospital of the RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, 1959 North East Pacific Avenue, Box 356521, Room BB1269, Seattle, WA 98195-6521, USA
| | - Marcus J Moeller
- Nephrology and Clinical Immunology, University Hospital of the RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| |
Collapse
|
94
|
Anders HJ, Fogo AB. Immunopathology of lupus nephritis. Semin Immunopathol 2014; 36:443-59. [PMID: 24402709 DOI: 10.1007/s00281-013-0413-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/02/2013] [Indexed: 01/04/2023]
Abstract
When patients with systemic lupus erythematosus (SLE) present with urinary abnormalities, a renal biopsy is usually needed to rule out or confirm lupus nephritis. Renal biopsy is also needed to define the type of renal manifestation as different entities are associated with different outcomes; hence, renal biopsy results shape lupus management. But why does lupus nephritis come in different shapes? Why do patients with SLE often show change over time in class of lupus nephritis or have mixed forms? How does autoimmunity in SLE evolve? Why does loss of tolerance against nuclear antigens preferentially affect the kidney? Why are immune complex deposits in different glomerular compartments associated with different outcomes? What determines crescent formation in lupus? In this review, we discuss these questions by linking the latest information on lupus pathogenesis into the context of the different classes of lupus nephritis. This should help the basic scientist, the pathologist, and the clinician to gain a more conceptual view on the immunopathology of lupus nephritis.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ziemssenstr. 1, 80336, Munich, Germany,
| | | |
Collapse
|
95
|
Holderied A, Anders HJ. Animal models of kidney inflammation in translational medicine. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.ddmod.2014.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
96
|
Liapis H, Romagnani P, Anders HJ. New insights into the pathology of podocyte loss: mitotic catastrophe. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1364-1374. [PMID: 24007883 DOI: 10.1016/j.ajpath.2013.06.033] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/23/2013] [Accepted: 06/25/2013] [Indexed: 12/24/2022]
Abstract
Podocytes represent an essential component of the kidney's glomerular filtration barrier. They stay attached to the glomerular basement membrane via integrin interactions that support the capillary wall to withstand the pulsating filtration pressure. Podocyte structure is maintained by a dynamic actin cytoskeleton. Terminal differentiation is coupled with permanent exit from the cell cycle and arrest in a postmitotic state. Postmitotic podocytes do not have an infinite life span; in fact, physiologic loss in the urine is documented. Proteinuria and other injuries accelerate podocyte loss or induce death. Mature podocytes are unable to replicate and maintain their actin cytoskeleton simultaneously. By the end of mitosis, cytoskeletal actin forms part of the contractile ring, rendering a round shape to podocytes. Therefore, when podocyte mitosis is attempted, it may lead to aberrant mitosis (ie, mitotic catastrophe). Mitotic catastrophe implies that mitotic podocytes eventually detach or die; this is a previously unrecognized form of podocyte loss and a compensatory mechanism for podocyte hypertrophy that relies on post-G1-phase cell cycle arrest. In contrast, local podocyte progenitors (parietal epithelial cells) exhibit a simple actin cytoskeleton structure and can easily undergo mitosis, supporting podocyte regeneration. In this review we provide an appraisal of the in situ pathology of mitotic catastrophe compared with other proposed types of podocyte death and put experimental and renal biopsy data in a unified perspective.
Collapse
Affiliation(s)
- Helen Liapis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri; Department of Internal Medicine (Renal), Washington University School of Medicine, St. Louis, Missouri.
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of de Novo Therapies (DENOTHE), Florence, Italy; Pediatric Nephrology Unit, Meyer Children's Hospital, Florence, Italy
| | - Hans-Joachim Anders
- Nephrology Center, Medical Hospital and Health Center IV, University of Munich Clinical Center-LMU, Campus Innenstadt, Munich, Germany
| |
Collapse
|